• No results found

Lipopolysaccharide restricts murine norovirus infection in macrophages mainly through NF-kB and JAK-STAT signaling pathway

N/A
N/A
Protected

Academic year: 2021

Share "Lipopolysaccharide restricts murine norovirus infection in macrophages mainly through NF-kB and JAK-STAT signaling pathway"

Copied!
13
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Contents lists available atScienceDirect

Virology

journal homepage:www.elsevier.com/locate/virology

Lipopolysaccharide restricts murine norovirus infection in macrophages

mainly through NF-kB and JAK-STAT signaling pathway

Peifa Yu, Yang Li, Yining Wang, Maikel P. Peppelenbosch, Qiuwei Pan

∗ Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands

A R T I C L E I N F O Keywords: Norovirus Inflammasome Interferon Lipopolysaccharide Macrophages A B S T R A C T

The inflammasome machinery has recently been recognized as an emerging pillar of innate immunity. However, little is known regarding the interaction between the classical interferon (IFN) response and inflammasome activation in response to norovirus infection. We found that murine norovirus (MNV-1) infection induces the transcription of IL-1β, a hallmark of inflammasome activation, which is further increased by inhibition of IFN response, but fails to trigger the release of mature IL-1β. Interestingly, pharmacological inflammasome inhibitors do not affect viral replication, but slightly reverse the inflammasome activator lipopolysaccharide (LPS)-medi-ated inhibition of MNV replication. LPS efficiently stimulates the transcription of IFN-β through NF-ĸB, which requires the transcription factors IRF3 and IRF7. This activates downstream antiviral IFN-stimulated genes (ISGs) via the JAK-STAT pathway. Moreover, inhibition of NF-ĸB and JAK-STAT signaling partially reverse LPS-mediated anti-MNV activity, suggesting additional antiviral mechanisms activated by NF-ĸB. This study reveals additional insight in host defense against MNV infection.

1. Introduction

Norovirus is a major cause of epidemic nonbacterial gastroenteritis worldwide. Currently, no vaccination or specific antiviral treatment is available except for supportive care and supplementation of fluids, exerting a great global economic burden and significant impact on public health (Karst et al., 2014). The lack of a robust human norovirus (HuNV) cell culture system is a key bottleneck for norovirus research. Recent studies have achieved some success in culturing HuNV in in-testinal enteroid or B cells with modest viral replication level (Ettayebi et al., 2016;Jones et al., 2015), and in zebrafish larvae (Van Dycke et al., 2019) and human induced pluripotent stem cell-derived in-testinal epithelial cells (Sato et al., 2019), but these systems have not been widely used. A HuNV subgenomic replicon model in Huh7 cell line is widely used, for instance for antiviral drug evaluation (Chang et al., 2006). Murine norovirus (MNV), permissive for replication in both cell culture and small-animal models, shares similar structural, biochemical and genetic features with HuNV, and thus has become a useful model for studying norovirus biology and associated pathology (Wobus et al., 2004,2006).

Innate immune response provides thefirst line of defense against viral infection through the recognition of pathogen associated mole-cular patterns (PAMPs) by its pattern recognition receptors (PRRs)

including the recognized Toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-I like receptor (RLRs) and the cyclic GMP-AMP synthase (cGAS) (Wu and Chen, 2014). During viral replication, the accumulated nucleic acids within cells can be sensed by certain PRRs, and then ra-pidly activate the following intracellular transcription factors such as nuclear factor-ĸB (NF-ĸB) and IFN-regulatory factors (IRF3 and IRF7), leading to secretion of cytokines and chemokines including IFNs (Schneider et al., 2014). The released IFNs bind to their receptors on the cell surface, and in turn activate Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway and further induce transcription of hundreds of IFN-stimulated genes (ISGs). A subset of ISGs are ultimate antiviral effectors limiting viral replication and spread (Schoggins et al., 2011). More recently, the inflammasome signaling has also been reported to exert a central role in the regulation of gastrointestinal health and disease (Man, 2018). Inflammasomes are multiprotein complexes that induce downstream immune responses to specific pathogens and host cell damage (Chen and Ichinohe, 2015). Once activated, inflammasomes can induce the activation of caspases and release of mature interleukin-1β (IL-1β) and IL-18, and execute a form of inflammatory cell death known as pyroptosis (Chen and Ichinohe, 2015;Man, 2018).

Lipopolysaccharide (LPS) is an important structural component of the outer membrane of gram-negative bacteria, acting as a potent

https://doi.org/10.1016/j.virol.2020.04.010

Received 4 March 2020; Received in revised form 17 April 2020; Accepted 18 April 2020

Corresponding author. Department of Gastroenterology and Hepatology, Erasmus MC, Room Na-1005, 's-Gravendijkwal 230, NL-3015 CE, Rotterdam, the

Netherlands.

E-mail address:q.pan@erasmusmc.nl(Q. Pan).

Virology 546 (2020) 109–121

Available online 24 April 2020

0042-6822/ © 2020 The Author(s). Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

(2)

inflammasome activator. Upon LPS recognition, TLR4 signaling can be activated by undergoing oligomerization and recruiting its downstream adaptors through interactions with the TIR (Toll-interleukin-1 receptor) domains, which further induces the proinflammatory cytokine expres-sion and IFNs secretion through myeloid differentiation primary re-sponse gene 88 (MyD88)-dependent or MyD88-independent pathways (Lu et al., 2008). Moreover, LPS has been reported to associate with non-canonical inflammasome activation by binding with caspase 11 and then cleaving gasdermin D to induce pyroptotic cell death (Broz and Dixit, 2016).

Inflammasome activation is essential in controlling viral infection, such as rotavirus and influenza virus (Ichinohe et al., 2009;Zhu et al., 2017). Previous studies have reported that MNV-1 was rapidly cleared in wild-type mice (Karst et al., 2003), but persisted much longer in NLRP6-deficient mice (Wang et al., 2015), suggesting the potential involvement of inflammasome activation in norovirus infection. A re-cent study has revealed that MNV infection induced secretion of mature IL-1β by activating NLRP3 inflammasome in primary bone marrow-derived macrophages (BMDMs) that primed with TLR2 agonist, and in STAT1 deficient BMDMs that without TLR2 priming (Dubois et al., 2019). In this study, we investigated whether the IFN and inflamma-some machineries interact and collectively respond to norovirus infec-tion, and LPS-mediated anti-norovirus response.

2. Materials and methods 2.1. Reagents

Human IFN-α (Thermo Scientific, the Netherlands) and mouse IFN-γ (ab9922, Abcam) were dissolved in PBS. Lipopolysaccharide (LPS; L6529, Sigma) and adenosine 5-triphosphate disodium salt hydrate (ATP; A3377, Sigma) were dissolved in PBS. Stocks of JAK inhibitor 1 (Santa Cruz Biotech, CA), Bayer 11–7085 (Santa Cruz Biotech, CA), MCC950 (inh-mcc, Invivogen) and Ac-YVAD-cmK (inh-yvad, Invivogen) were dissolved in DMSO. Antibodies against phospho-STAT1 (Ser727) (#9177), phospho-STAT1 (#9172), MDA5 (D74E4, #5321) and IRF1 (D5E4, #8478), were purchased from Cell Signaling Technology. GBP2 antibody (11854-1-AP) was purchased from Proteintech. Rabbit polyclonal antisera to MNV NS1-2 (Baker, 2012) was kindly provided by Prof. Vernon K. Ward (School of Biomedical Sciences, University of Otago, New Zealand). β-actin antibody (#sc-47778) was purchased from Santa Cruz Biotechnology. IRDye® 800CW-conjugated goat anti-rabbit and goat anti-mouse IgGs (Li-Cor Bioscience, Lincoln, USA) were used as secondary antibodies, as appropriate.

2.2. Cell culture and viruses

RAW264.7 and J774A.1 cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Lonza Verviers, Belgium) supplemented with 10% (vol/vol) heat-inactivated fetal calf serum (FCS; Hyclone, Logan, UT, USA), 100 μg/mL of streptomycin, and 100 IU/mL of penicillin. Human Huh7 hepatocellular carcinoma cells harboring a genotype 1 HuNV replicon (HG23) were kindly provided by Dr. Kyeong-Ok Chang (Kansas State University) (Chang et al., 2006). A neomycin resistant gene was engineered into ORF2, conferring HG23 resistance to neo-mycin. Gentamicin (G418; Gibco) was added to HG23 culture medium at 1.5 mg/mL for selection before experimentation. Mouse embryonic fibroblast cells (MEFs) (WT, NF-κB−/−and IRF3/7−/−) were kindly

provided by Dr. Sanna M. Mäkelä (National Institute for Health and Welfare Viral Infections Unit, Helsinki, Finland) with the permission of Dr. Michael J. Gale and Dr. A. Hoffmann (Mäkelä et al., 2015). Wild-type (WT) MEFs and MEFs from STAT1−/− mice were generously provided by Prof. Andrea Kröger (Helmholtz Centre for Infection Re-search) (Nandakumar et al., 2013).

MNV-1 (murine norovirus strain MNV-1.CW1) was produced by inoculating the virus (kindly provided by Prof. Herbert Virgin,

Department of Pathology and Immunology, Washington University School of Medicine) into RAW264.7 cells (Wobus et al., 2004). The MNV-1 cultures were purified, aliquoted, and stored at −80 °C for all subsequent experiments. The MNV-1 stock was quantified three in-dependent times by the 50% tissue culture infective dose (TCID50). 2.3. TCID50

MNV-1 was quantified by TCID50 assay (Hwang et al., 2014). Briefly, 10-fold dilutions of MNV-1 were inoculated into RAW264.7 cells grown in 96-well tissue culture plate at 1000 cells/ well. The plate was incubated at 37 °C for another 5 days, followed by observing the cytopathic effect (CPE) of each well under a light scope. The TCID50 was calculated by using the Reed-Muench method. 2.4. MNV-1 infection in macrophages

To determine whether MNV-1 infection could activate the in-flammasome, RAW264.7 (1 × 105 cells/well) and J774A.1

(1 × 105cells/well) cells were plated at 12-well plate and were rested

for 24 h at 37 °C. Cells were then either left uninfected or infected with MNV-1 at indicated multiplicity of infection (MOI) for 1 h at 37 °C. The MNV-1 inoculum was removed and the cells were washed three times with phosphate-buffered saline (PBS), and fresh medium was added back onto cells. LPS and ATP treated cells were set as positive control. Total RNA and cell supernatant samples were prepared at indicated time and stored at−80 °C before use.

For MNV-1 antiviral assay, RAW264.7 and J774A.1 cells were either left untreated or treated with LPS, IFN-γ or combination with indicated compounds for 6 h before inoculation with MNV-1 at 1 MOI. After 1 h inoculation, the cells were washed with PBS for three times to remove the residual viruses, and control medium or medium supplemented with LPS or mouse IFN-γ was added back onto cells. After 24 h of in-fection, total RNA, protein and supernatant samples were collected from the cells for further analysis.

2.5. HuNV antiviral assay

Twenty-four hours before experimentation, HG23 cells were cul-tured in the medium without gentamicin. HG23 cells were seeded into 48-well tissue culture plates (2.5 × 104

cells per well), treated with different concentrations of LPS or human IFN-α. After 48 h of treat-ment, total RNA was extracted from cells, and HuNV replication was determined by qRT-PCR analysis.

2.6. Enzyme-linked immunosorbent assay (ELISA)

Supernatant was analyzed for cytokine IL-1β secretion by an ELISA kit (KMC0012; Invitrogen) according to manufacturer's instructions. The absorbance was measured at 450 nm in an automatic microplate reader. Results were calculated based on a standard curve.

2.7. Quantitative real-time polymerase chain reaction

Total RNA was isolated with a Macherey NucleoSpin RNA II Kit (Bioke, Leiden, The Netherlands) and quantified with a Nanodrop ND-1000 (Wilmington, DE, USA). During RNA isolation, DNase was added to remove genomic DNA according to the manufacturer's instructions. cDNA was synthesized from 500 ng of RNA using a cDNA synthesis kit (TaKaRa Bio, Inc., Shiga, Japan). The cDNA of all targeted genes transcript were quantified by SYBR-Green-based (Applied Biosystems) real-time PCR on the StepOnePlus™ System (Thermo Fisher Scientific Life Sciences) according to the manufacturer's instructions. Human glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and murine GAPDH genes were used as reference genes to normalize gene expres-sion. The relative expression of targeted gene was calculated as 2−ΔΔCT,

(3)

whereΔΔCT =ΔCTsample - ΔCTcontrol (ΔCT= CT[targeted gene]

-CT[GAPDH]). All primer sequences are listed inSupplementary Table 1 and Table 2.

2.8. Western blotting

Cultured cells were lysed in Laemmli sample buffer containing 0.1 M DTT and heated 5 min at 95 °C, then loaded onto a 10% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) gel, then proteins were electrophoretically transferred onto a poly-vinylidene difluoride (PVDF) membrane (pore size, 0.45 μM; Invitrogen) for 2 h with an electric current of 250 mA. Subsequently, the membrane was blocked with a mixture of 2.5 mL blocking buffer (Odyssey) and 2.5 mL PBS containing 0.05% Tween 20 for 1 h, followed by overnight incubation with primary antibodies (1:1000) at 4 °C. The membrane was washed 3 times and then incubated with IRDye-con-jugated secondary antibody (1:5000) for 1 h. After washing 3 times, protein bands were detected with the Odyssey 3.0 Infrared Imaging System (Li-Cor Biosciences).

2.9. MTT assay

Cells were seeded into 96-well tissue culture plates and cell viability was assessed by adding 10 mM 3-(4,5-dimethyl-2-thiazolyl)-2,5-di-phenyl-2H-tetrazolium bromide (MTT) (Sigma, Zwijndrecht, the Netherlands). After 3 h, the medium was replaced with 100μL of DMSO and was incubated at 37 °C for another 50 min. The absorbance at 490 nm was recorded on the microplate absorbance reader (Bio-Rad, CA, USA).

2.10. Statistical analysis

Data are presented as the mean ± SEM. Comparisons between groups were performed with Mann-Whitney test using GraphPad Prism 5.0 (GraphPad Software Inc., La Jolla, CA, USA). Differences were considered significant at a P value less than 0.05.

3. Results

3.1. MNV-1 infection did not induce the release of mature IL-1β, but increased its mRNA expression, which was further augmented by inhibition of IFN pathway in macrophages

To investigate whether MNV-1 can activate inflammasome, J774A.1 cells were used because of its permissiveness for MNV-1 in-fection and competence in inflammasome activation (Hirano et al., 2017;Wobus et al., 2004). Wefirst determined the expression of IL-1β and IL-18 as markers of inflammasome activation in J774A.1 cells treated with LPS (the component in the outer membrane of gram-ne-gative bacteria) and ATP, the potent activators for inflammasome ac-tivation. We found that under stimulation by LPS and ATP, the mRNA and protein levels of IL-1β were significantly increased (Fig. 1A–C), and IL-18 mRNA level was moderately elevated (Fig. 1A). Although the IL-1β mRNA level was modestly elevated in MNV-1 infected J774A.1 cells (Fig. 1B), no significant differences in IL-1β protein level were observed in the supernatant of these cells (Fig. 1C). We confirmed active viral infection by detecting the viral NS1/2 protein in J774A.1 cells (Fig. 1D). In another macrophage cell line RAW264.7 cells, the IL-1β and IL-18 mRNA levels were hardly changed upon MNV-1 infection (Supplementary Fig. 1). However, treatment with LPS and ATP also failed to induce the expression of these genes (Supplementary Figs. 1A and 1B), indicating possible defects of the inflammasome machinery in RAW264.7 cells, which is consistent with a previous study (He et al., 2015).

We next investigated whether MNV-1 infection has a role in LPS-induced IL-1β transcription. As expected, the IL-1β mRNA level was

further increased in MNV-1 infected J774A.1 cells by LPS and ATP stimulation (Fig. 1E), and this effect was also observed in LPS-pre-treated cells (Fig. 1F). A recent study reported that MNV infection sti-mulated IL-1β RNA transcription and protein release in STAT1 deficient BMDMs (Dubois et al., 2019). Thus, we blocked the JAK-STAT pathway by using JAK-STAT inhibitor (JAK inhibitor 1), and found that inhibi-tion of JAK-STAT pathway attenuated MNV-1 induced ISG transcripinhibi-tion (Fig. 1G), but increased MNV-1 RNA level (Fig. 1H) and viral infection-induced IL-1β mRNA transcription (Fig. 1I), which is in accordance with the recent study (Dubois et al., 2019).

3.2. LPS potently inhibits MNV-1 replication largely independent of inflammasome

Norovirus cohabits with bacteria in the intestine. Co-infection of MNV and bacteria in macrophages has been reported to reduce the viral load (Agnihothram et al., 2015;Li et al., 2016). Thus, we further in-vestigated the direct effects of bacteria-derived LPS on MNV-1 re-plication, and found that treatment with LPS potently inhibits viral replication in J774A.1 cells (Fig. 2A and B). However, this inhibitory effect on MNV-1 is only partially reversed by pharmacological in-flammasome inhibitors MCC950 and YVAD-cmk, which did not affect viral replication (Fig. 2A and B) but decreased the release of mature IL-1β triggered by LPS and ATP stimulation in J774A.1 cells (Fig. 2C).

We next evaluated the anti-MNV effect of LPS in RAW264.7 cells, which are incompetent of inflammasome activation (He et al., 2015). IFN-γ was used as a control. Pre-treatment with LPS potently inhibited viral RNA and NS1-2 protein levels in a dose-dependently manner (Fig. 2D). Similar effects were observed when cells were first infected with MNV-1 then subjected to treatment with LPS (Fig. 2E). Moreover, the viral titers were also decreased by LPS and IFN-γ pre-treatment (Fig. 2F).

These results urged us to investigate the antiviral effect of LPS on HuNV replication by using the Huh7-based HuNV replicon cells (HG23). After two days of treatment, LPS failed to but human IFN-α effectively restricted HuNV replication without triggering major cyto-toxicities (Fig. 3A). It is widely recognized that IFNs exert antiviral effects via induction of the transcription of ISGs through JAK-STAT signaling pathway. We further revealed that expression of type I IFNs (α and β) were not significantly induced by both LPS and IFN-α (Fig. 3B and C). However, IFN-α but not LPS triggered the expression and phosphorylation of STAT1 (Fig. 3D), and induction of ISGs in-cluding ISG15, IFIT1 and MX1 in HG23 cells (Fig. 3E). This is consistent with previousfindings that hepatic epithelia cells do not respond to LPS stimulation (MacParland et al., 2016). This previous study also showed that LPS suppresses IFN response in hepatocytes by upregulating ubi-quitin-like protease 18 (USP18) expression, a negative regulator of type I IFN signaling (MacParland et al., 2016). However, LPS stimulation did not affect USP18 gene transcription in our study (Fig. 3F).

These results have demonstrated that LPS potently inhibits MNV replication in macrophages, but has no significant effect on HuNV re-plication in HG23 cells. This is probably related to the intrinsic property that epithelial HG23 cells are nonresponsive to LPS stimulation. 3.3. LPS stimulates the transcription of IFN-β and a wide range of ISGs

Given the dispensability of inflammasome in anti-MNV activity of LPS, we explored other possible mechanisms. It is generally recognized that LPS stimulation of macrophages induces production of many cy-tokines and inflammatory mediators including IRF-1, IL-6 and IFNs (Beutler and Rietschel, 2003; Guha and Mackman, 2001). We found that LPS stimulated the expression of the viral RNA sensor MDA5 in RAW264.7 and J774A.1 cells (Fig. 4A and B), and the increased tran-scription of IFN-β in both cell lines (Fig. 4C and D), respectively. ISGs are the ultimate antiviral effectors induced by IFNs through the JAK-STAT pathway. We found the activation of JAK-JAK-STAT pathway by

(4)

(caption on next page)

(5)

detecting the increased expression of STAT1 at both the protein (Fig. 4A and B) and transcriptional levels (Fig. 4E and F), respectively. More-over, the transcription of a wide range of ISGs was also stimulated by LPS (Fig. 4E and F), and IFN-γ treatment also induced the transcription of many ISGs in both cell lines (Supplementary Fig. 2). The induction of several important genes were further confirmed by immunoblotting (Fig. 4A and B).

3.4. NF-ĸB signaling is important for LPS-induced IFN-β and ISG transcription

It has been reported that recognition of LPS by TLR4 activates downstream transcription factors NF-ĸB, leading to the transcription of IFNs and ISGs (Moynagh, 2005). Thus, we further investigated whether NF-ĸB signaling is involved in LPS-induced IFN-β and ISG transcription in macrophages. Treatment with Bay 11–7085, a pharmacological in-hibitor of NF-ĸB activation, largely inhibits LPS-triggered transcription of IFN-β (Fig. 5A and C) and most of the ISGs (Fig. 5B and D) in both macrophage cell lines. Moreover, this inhibitor also attenuated LPS-induced expression and phosphorylation of STAT1, as well as ISG ex-pression such as IRF1 and GBP2 (Fig. 5E and F). Similarly, some ISGs induced by IFN-γ were also affected by Bay 11–7085 treatment in both cell lines (Supplementary Fig. 3).

To further determine the essential role of NF-ĸB pathway in LPS-induced ISG expression, wild-type (WT) and NF-ĸB deficient (NF-ĸB−/ −) MEFs were used. In NF-ĸB−/−but not the WT cells, LPS failed to

induce the transcription of IFN-β (Fig. 5G). A similar ISG induction pattern was also observed in NF-ĸB−/−cells. All of the genes detected

such as IRF1, ISG15, MX1 and GBP2 were significantly induced by LPS in WT cells but not in NF-ĸB−/−cells (Fig. 5H). Moreover, transcription

of some ISGs induced by IFN-γ were largely decreased in NF-ĸB−/−

cells (Supplementary Fig. 4A). Taken together, thesefindings revealed that NF-ĸB pathway is important for LPS-induced IFN-β and ISG tran-scription.

3.5. IRF3 and IRF7 are important for LPS-induced IFN-β and ISG transcription

LPS as the ligand activates TLR4, which in turn phosphorylates downstream IRF3 and IRF7, leading to their nuclear translocation and induction of type I IFN genes and co-stimulatory molecules (Moynagh, 2005). Moreover, IRF3 and IRF7 have been reported to play an im-portant role in controlling MNV-1 replication (Thackray et al., 2012). Thus, by using WT and IRF3/7 deficient (IRF3/7−/−) MEFs, we found

that IFN-β was not induced by LPS in IRF3/7−/− cells (Fig. 6A).

Moreover, the detected genes induced by LPS in WT cells were reduced in IRF3/7−/−cells (Fig. 6B). Compared to the WT cells, transcription of ISG15, ISG20, MX1 and IFIT1 appeared to be unchanged, while tran-scription of other ISGs such as IRF1, MX2, GBP2 and GBP5 induced by IFN-γ was decreased in IRF3/7−/−cells (Supplementary Fig. 4B). These

results suggested that IRF3 and IRF7 are also important for LPS-induced IFN-β and ISG transcription.

3.6. LPS induces ISG transcription largely through activation of JAK-STAT pathway

The JAK-STAT signaling pathway has been shown to be essential for LPS-mediated gene expression in macrophages (Kovarik et al., 1998; Ohmori and Hamilton, 2001). Consistent with these results, we also observed that LPS induced the expression and phosphorylation of STAT1 in both RAW264.7 and J774A.1 cells, respectively (Fig. 7C and D). To determine whether ISG induction by LPS is through the activa-tion of STAT1, we used JAK-STAT inhibitor to pharmacologically block the JAK-STAT pathway. We found that JAK inhibitor 1 partially blocked LPS induced transcription of IFN-β in both RAW264.7 and J774A.1 cells (Fig. 7A and B), while both the expression and phos-phorylation of STAT1 triggered by LPS and IFN-γ were largely atte-nuated by this inhibitor (Fig. 7C and D). Next, we tested the mRNA level of some ISGs in LPS-stimulated macrophages treated with JAK inhibitor 1, and found that most of the LPS inducible genes were affected by JAK inhibitor 1 (Fig. 7E and F), without triggering major cytotoxicities (Supplementary Fig. 5D). In addition, the mRNA levels of these genes induced by IFN-γ were largely diminished by this inhibitor (Supplementary Figs. 5A and 5B).

To further confirm the ISG induction ability of LPS is partially de-pendent on the JAK-STAT pathway, we performed experiments with MEFs from WT and STAT1−/−mice to examine the effect of STAT1 on ISG expression (Fig. 7G and H). We found that the abundance of de-tected ISGs triggered by LPS were also largely decreased in STAT1−/− cells compared to those in WT cells (Fig. 7I). As a control, the tran-scription of IFN-γ induced ISGs was significantly blocked in STAT1−/−

cells (Supplementary Fig. 5C). Together, these results demonstrated that LPS activates ISG transcription largely through the activation of the JAK-STAT pathway.

3.7. The NF-ĸB cascade contributes to LPS-mediated inhibition of MNV-1 replication partially through the JAK-STAT pathway

In respect to the anti-MNV effects of LPS, we found that Bay 11–7085, the NF-ĸB signaling inhibitor, partially reversed LPS and IFN-γ-mediated antiviral response against MNV-1 infection in RAW264.7 cells (Fig. 8A and E). Similar results were also observed in J774A.1 cells (Fig. 8B). Interestingly, the inhibitor alone enhanced viral replication (Fig. 8A, B and 8E), suggesting a basal defense mechanism of NF-ĸB signaling against MNV-1 infection. Moreover, we also de-termined the role of JAK-STAT pathway in LPS-mediated antiviral ac-tivity and found that the anti-MNV acac-tivity of LPS was only moderately reversed, whereas the anti-MNV effect of IFN-γ was largely abolished by JAK inhibitor 1 (Fig. 8C–E). Together, these results have demonstrated that NF-ĸB cascade contributes to LPS-mediated control of MNV-1 re-plication partially through the JAK-STAT pathway.

4. Discussion

Innate immune response provides thefirst line of defense against viral infection. Studies have revealed that compared to wild-type mice, mice lacking either type I and type II IFN receptors or STAT1 have higher titers and succumb to lethal MNV infection (Karst et al., 2003; Thackray et al., 2012), indicating the importance of IFNs in controlling

Fig. 1. MNV-1 infection did not induce the release of mature IL-1β, but increased its mRNA level in macrophages. J774A.1 cells were untreated or treated with LPS (400 ng/μl) for 4 h and then ATP (5 mM) for 40 min, or left uninfected or infected with MNV-1 (MOI 1) for 24 h, and total RNA and cell culture supernatants were collected. mRNA levels of IL-1β (A and B) (n = 6) and IL-18 (A) (n = 6), and protein level of IL-1β (C) (n = 4) were analyzed by qRT-PCR and ELISA, respectively. Western blotting (D) was used to analyze the viral infection in J774A.1 cells at indicated times. (E) J774A.1 cells were uninfected or infected with MNV-1 (MOI MNV-1) for 20 h, then treated with LPS (400 ng/μl) for 4 h and ATP (5 mM) for 40 min mRNA level of IL-1β and cytotoxicity were analyzed by qRT-PCR (n = 8). (F) J774A.1 cells were untreated or treated with LPS (400 ng/μl) for 4 h then uninfected or infected with MNV-1 (MOI 1) for 24 h mRNA level of IL-1β and cytotoxicity were analyzed by qRT-PCR (n = 8). J774A.1 cells were untreated or treated with JAK inhibitor 1 (10μM) for 6 h then uninfected or infected with MNV-1 (MOI 1) for 24 h mRNA levels of indicated genes (G) (n = 6), MNV-1 RNA (H) (n = 6) and IL-1β mRNA (I) (n = 6) were analyzed by qRT-PCR, respectively. Data were normalized to the untreated control (CTR, set as 1). *P < 0.05; **P < 0.01; ***P < 0.001; ns, not significant. β-actin was used as a loading control.

(6)

Fig. 2. LPS potently inhibits MNV-1 replication largely independent of inflammasome. J774A.1 cells were treated with LPS (400 ng/μl), MCC950 (5 μM), YVAD-cmk (5μM), or combination for 6 h, then infected with MNV-1 (MOI 1) for 24 h. (A) The viral RNA, NS1/2 protein and cytotoxicity were analyzed by qRT-PCR (n = 6), western bloting, and MTT (n = 16) assays, respectively. (B) The viral titers were also analyzed by TCID50 assay (n = 4–6). (C) J774A.1 cells were untreated or treated with LPS (400 ng/μl), MCC950 (5 μM), YVAD-cmk (5 μM), or combination for 4 h and then ATP (5 mM) for 40 min. The cell culture supernatants were collected and used for analysis of IL-1β by ELISA assay (n = 6). (D) RAW264.7 cells were untreated or treated with different concentrations of LPS as indicated, or IFN-γ (100 U/ml) for 6 h, then infected with MNV-1 (MOI 1) for 24 h. The viral RNA, NS1/2 protein and cytotoxicity were analyzed by qRT-PCR (n = 6), western blotting and MTT (n = 8) assay, respectively. (E) RAW264.7 cells were infected with MNV-1 (MOI 1) for 1 h, then added fresh medium or medium supplemented with LPS at indicated concentrations or IFN-γ (100 U/ml) for 24 h. The viral RNA, NS1/2 protein and cytotoxicity were analyzed by qRT-PCR (n = 3 independent experiments with 2–3 replicates each), western blotting and MTT (n = 8) assays, respectively. (F) RAW264.7 cells were untreated or treated with different con-centrations of LPS as indicated, or IFN-γ (100 U/ml) for 6 h, then infected with MNV-1 (MOI 1) for 24 h. The viral titers were analyzed by TCID50 assay (n = 6). Data were normalized to the untreated control (CTR, set as 1). *P < 0.05; **P < 0.01; ns, not significant. ND, not determined. β-actin was used as a loading control.

(7)

norovirus infection. Besides IFNs, inflammasome signaling also has a central role in innate immune response against viral infections (Zhu et al., 2017). Previous evidence indicated the potential role of in-flammasome in norovirus replication (Wang et al., 2015). However, in this study, MNV-1 infection failed to adequately activate inflammasome activation in macrophages, although moderate activation of IL-1β transcription was observed in J774A.1 cells. Similar results have been reported that MNV failed to secrete IL-1β in WT BMDMs, but presented caspase-1 processing ability (Dubois et al., 2019). In in flammasome-incompetent RAW264.7 cells, which lack the expression of ASC, one of the components for inflammasome complex formation (He et al., 2015), MNV-1 infection appeared not to affect IL-1β transcription. A recent study reported that MNV infection induced mature IL-1β secretion in STAT1 deficient BMDMs (Dubois et al., 2019). Compared to MNV-1 infection alone, we also observed the increased IL-1β transcription level induced by MNV-1 with JAK-STAT pathway inhibition. Studies have reported the crosstalk between IFN response and inflammasome acti-vation (Kopitar-Jerala, 2017). Here we revealed that inhibition of

JAK-STAT pathway reduced MNV-1 infection triggered IFN response, but increased MNV-induced IL-1β transcription. Moreover, decreased IFN response by inhibition of JAK-STAT pathway resulted in increased viral replication, which might further contribute to the upregulation of IL-1β transcription. Thus, whether there is a potential crosstalk between IFN response and inflammasome activation in response to MNV infection needs to be further studied.

Norovirus and bacteria are cohabitants of the intestine. Gram-ne-gative bacteria produce a large amount of LPS, which are naturally potent activators of inflammasome. It has been reported that hepatitis B virus (HBV) infection inhibits LPS-induced inflammasome activation, although HBV alone does not activate inflammasome (Yu et al., 2017). Moreover, studies have revealed that MNV infection can induced ma-ture IL-1β release in primed BMDMs with TLR2 agonist (Pam3CSK4) and TLR4 agonist (LPS) (Dubois et al., 2019), and here we found that MNV-1 augmented LPS-induced IL-1β transcription. Interestingly, we observed that LPS inhibited MNV-1 replication, but this inhibitory ef-fect was only slightly reversed by inflammasome inhibitors, suggesting

Fig. 3. HuNV replication is not affected by LPS in the replicon model. (A) HG23 cells were untreated or treated with different concentrations of LPS as indicated, or IFN-α (200 IU/ml). After 2 days of treatment the antiviral activities and cytotoxicity were measured by qRT-PCR (n = 6) and MTT assay (n = 16), respectively. qRT-PCR analysis (n = 4) of mRNA levels of IFN-α (B), IFN-β (C), STAT1 (D upper panel), ISGs (E), USP18 (F) and western blotting analysis of STAT1 expression and phosphorylation (D lower panel) in HG23 cells untreated or treated with LPS (100 ng/ml) or IFN-α (200 IU/ml). Data were normalized to the untreated control (CTR, set as 1). *P < 0.05; **P < 0.01; ***P < 0.001; ns, not significant. β-actin was used as a loading control.

(8)

additional mechanisms contributing to the anti-MNV activity of LPS. These inhibitors might have a negative role on LPS-triggered expression of inflammatory factors, which could aid to LPS-mediated antiviral response against MNV infection to some extent. However, we do not exclude that there could be more active interactions of norovirus with

inflammasome in other cell types, which deserve to be further in-vestigated. Moreover, blocking NLRP3 inflammasome activation has been reported to reduce MNV-induced immunopathology in STAT1-deficient mice (Dubois et al., 2019). In addition, we found that in-flammasome inhibitors alone appear not to affect the viral replication,

Fig. 4. LPS stimulates the transcription of a wide range of ISGs. Western blotting analysis of ISG protein levels in RAW264.7 cells (A) and J774A.1 cells (B) untreated or treated with LPS (100 ng/ml) or IFN-γ (100 U/ml) for 6 h qRT-PCR analysis of mRNA level of IFNs (C and D) (n = 6) and ISGs (E and F) (n = 4) in RAW264.7 cells and J774A.1 cells that untreated or treated with LPS (100 ng/ml) for 6 h, respectively. Data were normalized to the untreated control (CTR, set as 1). *P < 0.05; **P < 0.01; ns, not significant. β-actin was used as a loading control.

(9)

Fig. 5. NF-ĸB signaling is important for LPS-induced IFN-β and ISG transcription. Cells were untreated or treated with LPS (100 ng/ ml), Bay 11–7085 (5 μM) or combination for 6 h qRT-PCR analysis of IFN-β (A) and ISG (B) mRNA levels in RAW264.7 cells (n = 6). qRT-PCR analysis of IFN-β (C) and ISG (D) mRNA levels in J774A.1 cells (n = 6). Western blot-ting analysis of STAT1 expression and phos-phorylation, and expression of IRF1 and GBP2 in RAW264.7 cells (E) and J774A.1 cells (F) untreated or treated with LPS (100 ng/ml), IFN-γ (100 U/ml), Bay 11–7085 (5 μM) or combination for 6 h qRT-PCR analysis of mRNA levels of IFN-β (G) (n = 6) and ISGs (H) (n = 6) in WT and NF-ĸB−/− MEFs untreated or treated with LPS (100 ng/ml) for 6 h. Data in (A, B, C, D and G) were normalized to the untreated control (CTR, set as 1). Data in (H) were normalized to untreated WT and NF-ĸB−/− MEFs, respectively (both set as 1). *P < 0.05; **P < 0.01.β-actin was used as a loading control.

(10)

and similar results were also demonstrated that MNV replication rates were not affected in WT or NLRP3−/−BMDMs (Dubois et al., 2019).

As a type of PRRs, TLRs are responsible for the recognition of in-fectious agents leading to activation of innate immune response (Hertzog et al., 2003). It has been reported that TLR7 activation by its agonists displays potent antiviral effects against norovirus infection by stimulating innate immune response genes (Tuipulotu et al., 2018). As a ligand of TLR4, LPS can activate transcription factors NF-ĸB and IRFs, leading to the activation of IFN signaling (Moynagh, 2005). IFNs are central to the innate control of MNV infection (Changotra et al., 2009; Hwang et al., 2012;Nice et al., 2015). Here we found that LPS potently induces IFN-β transcription, and pharmacological inhibition of NF-ĸB signaling significantly blunts LPS-triggered IFN-β transcription, which is further confirmed in NF-ĸB deficient cells. Moreover, transcription factors IRF3 and IRF7 can regulate IFN-mediated inhibition of MNV-1 replication (Thackray et al., 2012), and we found in this study that IFN-β transcription induced by LPS is largely attenuated in IRF3 and IRF7 deficient cells.

ISGs are regarded as the ultimate antiviral effectors, and several ISGs such as IRF1, ISG15 and GBP2 have been reported to mediate the anti-norovirus effect of IFN (Biering et al., 2017;Maloney et al., 2012; Orchard et al., 2019;Rodriguez et al., 2014). In this study, we found that LPS also potently triggers ISG expression via the transcription factors including NF-ĸB, IRF3 and IRF7. Moreover, blocking NF-ĸB signaling attenuates LPS and IFN-γ mediated control of MNV-1 re-plication. JAK-STAT pathway is essential for IFN-induced ISG expres-sion (Ohmori and Hamilton, 2001;Shuai and Liu, 2003) and host innate immunity against MNV infection (Karst et al., 2003). We revealed that induction of ISGs triggered by LPS or IFN-γ are dramatically decreased by inhibition of the JAK-STAT pathway, which also largely abolishes IFN-γ mediated inhibition for MNV-1 replication. However, blocking this pathway partially reverses LPS-mediated anti-MNV ability. This indicates that LPS-triggered ISGs are partially responsible for its anti-viral response against MNV-1 infection in macrophages, and other un-known antiviral mechanisms mediated by LPS may be involved. Fur-thermore, the non-canonical mechanisms regulating ISG transcription have been reported (Wang et al., 2017). Induction of ISGs via NF-kB cascade might do not require JAK-STAT signaling, and this may account for the partial restoration of MNV replication by the inhibitors.

Accumulating evidences have demonstrated the interaction between gut microbiota and intestinal viruses including norovirus (Baldridge

et al., 2015;Jones et al., 2014;Miura et al., 2013). Several mechanisms have been reported on how intestinal viruses interact with bacteria. On one hand, gut microbiota has been revealed to facilitate MNV replica-tion through an antagonistic mechanism to IFN-lambda in the intestine (Baldridge et al., 2015), and prompt HuNV infection in intestinal B cells (Jones et al., 2014). Bacterial component of the enteric microbiome plays an essential role in the establishment of viral persistent infection. For instance, histo-blood group antigen (HBGA)-like substances im-prove virion stability and protect HuNV from acute heat stress (Li et al., 2015), and infection of B cells (Jones et al., 2014). It has been also reported that LPS binding promotes viral infectivity by enhancing virion stability and cell attachment (Kuss et al., 2011;Robinson et al., 2014), but fails to enhance HuNV infection of B cells in vitro (Jones et al., 2014), and inhibits MNV-1 replication in macrophages in this study. On the other hand, previous studies have presented the antag-onistic mechanisms that Enterobacter cloacae, a member of enteric commensal bacteria specifically binds HuNV, and inhibits virus in-fectivity in gnotobiotic pigs by enhancing the gut immunity (Lei et al., 2016). In addition, the microbiota-host interaction can also modulate the production of immune system molecules, resulting in anti-MNV effects. Thus, the interactions between intestinal bacteria and viruses collectively modulate the infection outcomes.

In summary, our study demonstrates that MNV-1 moderately trig-gers IL-1β transcription, and is augmented by inhibition of IFN pathway in macrophages. Importantly, LPS potently inhibits MNV-1 replication, might through inflammasome to some extent but mainly via the NF-ĸB cascade. Activation of NF-ĸB by LPS drives the expression of IFN-β through IRF3 and IRF7. This activates the downstream antiviral ISGs via the JAK-STAT pathway. Moreover, inhibition of NF-ĸB and JAK-STAT signaling partially reversed the anti-MNV activity of LPS, sug-gesting additional antiviral mechanisms activated by NF-ĸB, which need to be further studied. Our study has revealed new insight in virus-cell interactions, and may provide rationale for new therapeutic de-velopment against norovirus infection.

CRediT authorship contribution statement

Peifa Yu: Conceptualization, Methodology, Investigation, Writing -original draft, Writing - review & editing.Yang Li: Methodology, Data curation, Writing - review & editing.Yining Wang: Methodology, Data curation, Writing - review & editing. Maikel P. Peppelenbosch:

Fig. 6. IRF3 and IRF7 are important for LPS-induced IFN-β and ISG transcription. qRT-PCR analysis of mRNA levels of IFN-β (A) (n = 6) and ISGs (B) (n = 6) in WT and IRF3/7−/− MEFs untreated or treated with LPS (100 ng/ml) for 6 h. Data in (A) were normalized to the untreated control (CTR, set as 1). Data in (B) were normalized to untreated WT and IRF3/7−/− MEFs, respectively (both set as 1). **P < 0.01.

(11)

Fig. 7. LPS induces ISG transcription largely through activation of JAK-STAT pathway. qRT-PCR analysis of IFN-β mRNA levels in RAW264.7 cells (A) and J774A.1 cells (B) untreated or treated with LPS (100 ng/ml) or JAK inhibitor 1 (10μM) for 6 h (n = 4). Western blotting analysis of STAT1 expression and phosphorylation in RAW264.7 cells (C) and J774A.1 cells (D) untreated or treated with LPS (100 ng/ml), IFN-γ (100 U/ml), JAK inhibitor 1 (10 μM) or combination for 6 h qRT-PCR analysis of ISG mRNA levels in RAW264.7 cells (E) and J774A.1 cells (F) untreated or treated with LPS (100 ng/ml), IFN-γ (100 U/ml), JAK inhibitor 1 (10μM) or combination for 6 h (n = 4). WT and STAT1−/−MEFs were analyzed to determine the relative abundance of STAT1 mRNA level (G) by qRT-PCR

(n = 6) and protein level (H) by western blotting. (I) qRT-PCR analysis of ISG mRNA levels in WT and STAT1−/−MEFs untreated or treated with LPS (100 ng/ml) for 6 h (n = 6). Data in (A, B, E, F and G) were normalized to untreated control (CTR, set as 1). Data in (I) were normalized to untreated WT and STAT1−/−MEFs, respectively (both set as 1). *P < 0.05; **P < 0.01.β-actin was used as a loading control.

(12)

Conceptualization, Supervision, Writing - review & editing. Qiuwei Pan: Conceptualization, Methodology, Resources, Supervision, Writing - review & editing.

Declaration of competing interest

The authors declare that there are no conflicts of interest. Acknowledgements

We gratefully acknowledge Prof. Herbert W. Virgin (Washington University, St Louis, MO, USA) for providing us the MNV-1; Dr. Kyeong-Ok Chang (Kansas State University, USA) for providing the HuNV re-plicon; Prof. Vernon K. Ward (School of Biomedical Sciences, University of Otago, New Zealand) for providing rabbit polyclonal antisera to MNV NS1-2; Dr. Sanna M. Mäkelä (National Institute for Health and Welfare Viral Infections Unit, Helsinki, Finland) for providing the WT, NF-κB−/ −and IRF3/7−/−MEFs; and Prof. Andrea Kröger (Helmholtz Centre for

Infection Research) for providing the WT and STAT1−/−MEFs. This research is supported by the China Scholarship Council for funding PhD fellowships to P. Yu (No. 201708620177), Y. Li (No. 201703250073) and Y. Wang (No. 201903250082).

Appendix A. Supplementary data

Supplementary data to this article can be found online athttps:// doi.org/10.1016/j.virol.2020.04.010.

References

Agnihothram, S.S., Basco, M.D.S., Mullis, L., Foley, S.L., Hart, M.E., Sung, K., Azevedo, M.P., 2015. Infection of murine macrophages by Salmonella enterica serovar Heidelberg blocks murine norovirus infectivity and virus-induced apoptosis. PloS One 10, e0144911.

Baker, E.S., 2012. Characterisation of the NS1-2 and NS4 Proteins of Murine Norovirus. University of Otago.

Baldridge, M.T., Nice, T.J., McCune, B.T., Yokoyama, C.C., Kambal, A., Wheadon, M., Diamond, M.S., Ivanova, Y., Artyomov, M., Virgin, H.W., 2015. Commensal microbes and interferon-λ determine persistence of enteric murine norovirus infection. Science 347, 266–269.

Beutler, B., Rietschel, E.T., 2003. Innate immune sensing and its roots: the story of en-dotoxin. Nat. Rev. Immunol. 3, 169–176.

Biering, S.B., Choi, J., Halstrom, R.A., Brown, H.M., Beatty, W.L., Lee, S., McCune, B.T., Dominici, E., Williams, L.E., Orchard, R.C., 2017. Viral replication complexes are targeted by LC3-guided interferon-inducible GTPases. Cell Host Microbe 22, 74–85 e77.

Broz, P., Dixit, V.M., 2016. Inflammasomes: mechanism of assembly, regulation and signalling. Nat. Rev. Immunol. 16, 407–420.

Chang, K.O., Sosnovtsev, S.V., Belliot, G., King, A.D., Green, K.Y., 2006. Stable expression of a Norwalk virus RNA replicon in a human hepatoma cell line. Virology 353, 463–473.

Changotra, H., Jia, Y., Moore, T.N., Liu, G., Kahan, S.M., Sosnovtsev, S.V., Karst, S.M., 2009. Type I and type II interferons inhibit the translation of murine norovirus proteins. J. Virol. 83, 5683–5692.

Chen, I.Y., Ichinohe, T., 2015. Response of host inflammasomes to viral infection. Trends Microbiol. 23, 55–63.

Dubois, H., Sorgeloos, F., Sarvestani, S.T., Martens, L., Saeys, Y., Mackenzie, J.M., Lamkanfi, M., van Loo, G., Goodfellow, I., Wullaert, A., 2019. Nlrp3 inflammasome activation and Gasdermin D-driven pyroptosis are immunopathogenic upon gastro-intestinal norovirus infection. PLoS Pathog. 15, e1007709.

Ettayebi, K., Crawford, S.E., Murakami, K., Broughman, J.R., Karandikar, U., Tenge, V.R., Neill, F.H., Blutt, S.E., Zeng, X.L., Qu, L., 2016. Replication of human noroviruses in stem cell–derived human enteroids. Science 353, 1387–1393.

Guha, M., Mackman, N., 2001. LPS induction of gene expression in human monocytes. Cell. Signal. 13, 85–94.

Fig. 8. The NF-ĸB cascade contributes to LPS-mediated inhibition of MNV-1 replication partially through the JAK-STAT pathway. qRT-PCR (n = 6) and western blotting analysis of MNV-1 viral RNA and protein levels in RAW264.7 cells (A) and J774A.1 cells (B) untreated or treated with LPS (100 ng/ml), IFN-γ (100 U/ml), Bay 11–7085 (5 μM) or combination for 6 h, then infected with MNV-1 (MOI 1) for 24 h qRT-PCR (n = 6) and western blotting analysis of MNV-1 viral RNA and protein levels in RAW264.7 cells (C) and J774A.1 cells (D) untreated or treated with LPS (100 ng/ml), IFN-γ (100 U/ ml), JAK inhibitor 1 (10μM) or combination for 6 h, then infected with MNV-1 (MOI 1) for 24 h. (E) The viral titers were also analyzed by TCID50 assay (n = 4–9). Data were normalized to the untreated control (CTR, set as 1). *P < 0.05; **P < 0.01.β-actin was used as a loading control.

(13)

He, W.T., Wan, H., Hu, L., Chen, P., Wang, X., Huang, Z., Yang, Z.H., Zhong, C.Q., Han, J., 2015. Gasdermin D is an executor of pyroptosis and required for interleukin-1β se-cretion. Cell Res. 25, 1285.

Hertzog, P.J., O'Neill, L.A., Hamilton, J.A., 2003. The interferon in TLR signaling: more than just antiviral. Trends Immunol. 24, 534–539.

Hirano, S., Zhou, Q., Furuyama, A., Kanno, S., 2017. Differential regulation of IL-1β and IL-6 release in murine macrophages. Inflammation 40, 1933–1943.

Hwang, S., Alhatlani, B., Arias, A., Caddy, S.L., Christodoulou, C., Bragazzi Cunha, J., Emmott, E., Gonzalez‐Hernandez, M., Kolawole, A., Lu, J., 2014. Murine norovirus: propagation, quantification, and genetic manipulation. Curr. Protoc. Microbiol. 33 15K-2.

Hwang, S., Maloney, N.S., Bruinsma, M.W., Goel, G., Duan, E., Zhang, L., Shrestha, B., Diamond, M.S., Dani, A., Sosnovtsev, S.V., 2012. Nondegradative role of Atg5-Atg12/ Atg16L1 autophagy protein complex in antiviral activity of interferon gamma. Cell Host Microbe 11, 397–409.

Ichinohe, T., Lee, H.K., Ogura, Y., Flavell, R., Iwasaki, A., 2009. Inflammasome re-cognition of influenza virus is essential for adaptive immune responses. J. Exp. Med. 206, 79–87.

Jones, M.K., Grau, K.R., Costantini, V., Kolawole, A.O., De Graaf, M., Freiden, P., Graves, C.L., Koopmans, M., Wallet, S.M., Tibbetts, S.A., 2015. Human norovirus culture in B cells. Nat. Protoc. 10, 1939–1947.

Jones, M.K., Watanabe, M., Zhu, S., Graves, C.L., Keyes, L.R., Grau, K.R., Gonzalez-Hernandez, M.B., Iovine, N.M., Wobus, C.E., Vinjé, J., 2014. Enteric bacteria promote human and mouse norovirus infection of B cells. Science 346, 755–759.

Karst, S.M., Wobus, C.E., Goodfellow, I.G., Green, K.Y., Virgin, H.W., 2014. Advances in norovirus biology. Cell Host Microbe 15, 668–680.

Karst, S.M., Wobus, C.E., Lay, M., Davidson, J., Virgin, H.W., 2003. STAT1-dependent innate immunity to a Norwalk-like virus. Science 299, 1575–1578.

Kopitar-Jerala, N., 2017. The role of interferons in inflammation and inflammasome ac-tivation. Front. Immunol. 8, 873.

Kovarik, P., Stoiber, D., Novy, M., Decker, T., 1998. Stat1 combines signals derived from IFN‐γ and LPS receptors during macrophage activation. EMBO J. 17, 3660–3668. Kuss, S.K., Best, G.T., Etheredge, C.A., Pruijssers, A.J., Frierson, J.M., Hooper, L.V.,

Dermody, T.S., Pfeiffer, J.K., 2011. Intestinal microbiota promote enteric virus re-plication and systemic pathogenesis. Science 334, 249–252.

Lei, S., Samuel, H., Twitchell, E., Bui, T., Ramesh, A., Wen, K., Weiss, M., Li, G., Yang, X., Jiang, X., 2016. Enterobacter cloacae inhibits human norovirus infectivity in gno-tobiotic pigs. Sci. Rep. 6, 25017.

Li, D., Breiman, A., Le Pendu, J., Uyttendaele, M., 2015. Binding to histo-blood group antigen-expressing bacteria protects human norovirus from acute heat stress. Front. Microbiol. 6, 659.

Li, D., Breiman, A., Le Pendu, J., Uyttendaele, M., 2016. Anti-viral effect of bifido-bacterium adolescentis against noroviruses. Front. Microbiol. 7, 864.

Lu, Y.C., Yeh, W.C., Ohashi, P.S., 2008. LPS/TLR4 signal transduction pathway. Cytokine 42, 145–151.

MacParland, S.A., Ma, X.Z., Chen, L., Khattar, R., Cherepanov, V., Selzner, M., Feld, J.J., Selzner, N., McGilvray, I.D., 2016. LPS and TNF-α inhibit interferon-signaling in hepatocytes by increasing USP18 expression. J. Virol. 90, 5549–5560. Mäkelä, S.M., Österlund, P., Westenius, V., Latvala, S., Diamond, M.S., Gale, M.,

Julkunen, I., 2015. RIG-I signaling is essential for influenza B virus-induced rapid interferon gene expression. J. Virol. 89, 12014–12025.

Maloney, N.S., Thackray, L.B., Goel, G., Hwang, S., Duan, E., Vachharajani, P., Xavier, R., Virgin, H.W., 2012. Essential cell autonomous role for interferon regulatory factor 1 in interferon-γ-mediated inhibition of norovirus replication in macrophages. J. Virol. 86, 12655–12664.

Man, S.M., 2018. Inflammasomes in the gastrointestinal tract: infection, cancer and gut microbiota homeostasis. Nat. Rev. Gastroenterol. Hepatol. 1, 721–737.

Miura, T., Sano, D., Suenaga, A., Yoshimura, T., Fuzawa, M., Nakagomi, T., Nakagomi, O., Okabe, S., 2013. Histo-blood group antigen-like substances of human enteric bacteria as specific adsorbents for human noroviruses. J. Virol. 87, 9441–9451.

Moynagh, P.N., 2005. TLR signalling and activation of IRFs: revisiting old friends from the NF-κB pathway. Trends Immunol. 26, 469–476.

Nandakumar, R., Finsterbusch, K., Lipps, C., Neumann, B., Grashoff, M., Nair, S., Hochnadel, I., Lienenklaus, S., Wappler, I., Steinmann, E., 2013. Hepatitis C virus replication in mouse cells is restricted by IFN-dependent and-independent mechan-isms. Gastroenterology 145, 1414–1423 e1411.

Nice, T.J., Baldridge, M.T., McCune, B.T., Norman, J.M., Lazear, H.M., Artyomov, M., Diamond, M.S., Virgin, H.W., 2015. Interferon-λ cures persistent murine norovirus infection in the absence of adaptive immunity. Science 347, 269–273.

Ohmori, Y., Hamilton, T.A., 2001. Requirement for STAT1 in LPS‐induced gene expres-sion in macrophages. J. Leukoc. Biol. 69, 598–604.

Orchard, R.C., Sullender, M.E., Dunlap, B.F., Balce, D.R., Doench, J.G., Virgin, H.W., 2019. Identification of antinorovirus genes in human cells using genome-wide CRISPR activation screening. J. Virol. 93 e01324-01318.

Robinson, C.M., Jesudhasan, P.R., Pfeiffer, J.K., 2014. Bacterial lipopolysaccharide binding enhances virion stability and promotes environmentalfitness of an enteric virus. Cell Host Microbe 15, 36–46.

Rodriguez, M.R., Monte, K., Thackray, L.B., Lenschow, D.J., 2014. ISG15 functions as an interferon-mediated antiviral effector early in the murine norovirus life cycle. J. Virol. 88, 9277–9286.

Sato, S., Hisaie, K., Kurokawa, S., Suzuki, A., Sakon, N., Uchida, Y., Yuki, Y., Kiyono, H., 2019. Human norovirus propagation in human induced pluripotent stem cell-derived intestinal epithelial cells. Cell Mol. Gastroenterol. Hepatol. 7, 686–688 e685. Schneider, W.M., Chevillotte, M.D., Rice, C.M., 2014. Interferon-stimulated genes: a

complex web of host defenses. Annu. Rev. Immunol. 32, 513–545.

Schoggins, J.W., Wilson, S.J., Panis, M., Murphy, M.Y., Jones, C.T., Bieniasz, P., Rice, C.M., 2011. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature 472, 481–485.

Shuai, K., Liu, B., 2003. Regulation of JAK–STAT signalling in the immune system. Nat. Rev. Immunol. 3, 900–911.

Thackray, L.B., Duan, E., Lazear, H.M., Kambal, A., Schreiber, R.D., Diamond, M.S., Virgin, H.W., 2012. Critical role for IRF-3 and IRF-7 in type I interferon-mediated control of murine norovirus replication. J. Virol. 86, 13515–13523.

Tuipulotu, D.E., Netzler, N.E., Lun, J.H., Mackenzie, J.M., White, P.A., 2018. TLR7 ago-nists display potent antiviral effects against norovirus infection via innate stimula-tion. Antimicrob. Agents Chemother., AAC. 62, e02417–17.

Van Dycke, J., Ny, A., Conceicao-Neto, N., Maes, J., Hosmillo, M., Cuvry, A., Goodfellow, I., Nogueira, T.C., Verbeken, E., Matthijnssens, J., de Witte, P., Neyts, J., Rocha-Pereira, J., 2019. A robust human norovirus replication model in zebrafish larvae. PLoS Pathog. 15, e1008009.

Wang, P., Zhu, S., Yang, L., Cui, S., Pan, W., Jackson, R., Zheng, Y., Rongvaux, A., Sun, Q., Yang, G., 2015. Nlrp6 regulates intestinal antiviral innate immunity. Science 350, 826–830.

Wang, W., Xu, L., Su, J., Peppelenbosch, M.P., Pan, Q., 2017. Transcriptional regulation of antiviral interferon-stimulated genes. Trends Microbiol. 25, 573–584.

Wobus, C.E., Karst, S.M., Thackray, L.B., Chang, K.-O., Sosnovtsev, S.V., Belliot, G., Krug, A., Mackenzie, J.M., Green, K.Y., Virgin Iv, H.W., 2004. Replication of Norovirus in cell culture reveals a tropism for dendritic cells and macrophages. PLoS Biol. 2, e432. Wobus, C.E., Thackray, L.B., Virgin, H.W., 2006. Murine norovirus: a model system to

study norovirus biology and pathogenesis. J. Virol. 80, 5104–5112.

Wu, J., Chen, Z.J., 2014. Innate immune sensing and signaling of cytosolic nucleic acids. Annu. Rev. Immunol. 32, 461–488.

Yu, X., Lan, P., Hou, X., Han, Q., Lu, N., Li, T., Jiao, C., Zhang, J., Zhang, C., Tian, Z., 2017. HBV inhibits LPS-induced NLRP3 inflammasome activation and IL-1β pro-duction via suppressing the NF-κB pathway and ROS propro-duction. J. Hepatol. 66, 693–702.

Zhu, S., Ding, S., Wang, P., Wei, Z., Pan, W., Palm, N.W., Yang, Y., Yu, H., Li, H.-B., Wang, G., 2017. Nlrp9b inflammasome restricts rotavirus infection in intestinal epithelial cells. Nature 546, 667.

Referenties

GERELATEERDE DOCUMENTEN

The Resilience Score Card, based on Aldrich’s research on recovery, resilience, and social capital, showed the level of social capital of different neighbourhoods

In contrast to observations for pathogens such as Sal- monella and Mycobacterium, we found that strains belong- ing to different Bcc species, causing either acute fatal

Na die sondeval word sy kultuurskeppende taak duideliker ~mlyn en ui tgebrei ('rgl. die skepping van kultuur, wat inderdaad impliseer die toepassing van Goddelike

Salmonella associations in control embryos compared with the irf8 knockdown groups, supporting that the Lc3 response observed is mainly driven by macrophages ( Figure 4(e,f)

Our data link Hif-1a to proinflammatory macrophage Il-1b transcription in vivo during early mycobacterial infection and importantly highlight a host protective mechanism,

The method of PCTS for preclinical drug testing is highly useful due to several advantages: 1) preservation of the 3D structure and cellular organization, 2) reproducibility,

Deze bodembacteriën produceren bij het omzetten van het organische product giftige stoffen en daarmee kunnen vele schadelijke organismen in de bodem zoals aaltjes en

Sea Scrolls and Hellenistic-Early Roman Period Judaism, Studies on the Texts of the Desert of Judah 67 (Leiden: Brill, 2007); Jonathan Ben-Dov, Head of All Years: Astronomy